• 沒有找到結果。

視網膜色素上皮細胞轉殖α6β4integrin以增進黏著於細胞外間質─視網膜色素細移殖之前驅實驗

N/A
N/A
Protected

Academic year: 2021

Share "視網膜色素上皮細胞轉殖α6β4integrin以增進黏著於細胞外間質─視網膜色素細移殖之前驅實驗"

Copied!
39
0
0

加載中.... (立即查看全文)

全文

(1)

行政院國家科學委員會專題研究計畫 成果報告

視網膜色素上皮細胞轉殖α6β4integrin 以增進黏著於細

胞外間質─視網膜色素細移殖之前驅實驗

研究成果報告(精簡版)

計 畫 類 別 : 個別型 計 畫 編 號 : NSC 95-2314-B-002-159- 執 行 期 間 : 95 年 08 月 01 日至 96 年 07 月 31 日 執 行 單 位 : 國立臺灣大學醫學院眼科 計 畫 主 持 人 : 楊中美 共 同 主 持 人 : 楊長豪 計畫參與人員: 碩士級-專任助理:張鳳書 處 理 方 式 : 本計畫可公開查詢

中 華 民 國 96 年 10 月 26 日

(2)

Overexpression of

and Integrin Enhances Human Retinal

Pigment Epithelial Cells Adhesion and Proliferation on Layers of

Br

uc

h’

s

Me

mbr

ane

Chung-May Yang1, I-Mo Fang1, 2, Chang-Hao Yang1, Muh-Shy Chen1

From the1Department of Ophthalmology, National Taiwan University Hospital,

Taipei, Taiwan, and the2Department of Ophthalmology, Taipei City Hospital

Zhongxiao Branch, Taipei, Taiwan

Reprint requests to:

Chang-Hao Yang, MD, PhD

Department of Ophthalmology

National Taiwan University Hospital

No.7, Chung-Shan S. Rd. Taipei, Taiwan

TEL: 886-2-23123456 ext. 3193

Fax: 886-2-23412875

(3)

Abstract.

Purpose. To elucidate the roles of integrinon retinal pigment epithelial (RPE) cells adhesion and proliferation on extracellular matrix and to investigate whether

overexpression of integrinandcould promote adhesion and proliferation of RPE cells on Bruch’s membrane.

Mehtods. The expression of integrinmRNA and surface protein in ARPE-19 cells was analyzed by reverse transcription-polymerase chain reaction (RT-PCR) and

flow cytometry, respectively. Transfectants containing mutatedand

cDNAwhich were generated by site-directed mutagenesis, was used to test the adhesion on extracellular matrix. Mechanical and enzymatic techniques were used to

expose varied layers of Bruch’s membrane. The attachment of ARPE-19 cells

transfected with- and-cDNA on different layers of Bruch’s membrane was determined by adhesion analysis. Cell morphology and surface coverage were

evaluated by scanning electron microscopy.

Result. Integrin6and4mRNA and proteins were constitutionally expressed in

ARPE-19 cells. The adhesion rate of cells to laminin was decreased by cells

expressingmutant ormtant. The-cDNA transfectants displayed enhanced expression of the integrinandand increased adhesion to laminin, fibronectin and collagen type IV, whereas-cDNA transfectants showed increased expression of

(4)

theintegrin and enhanced attachment only to laminin. In Bruch’s membrane explant model,transfectants promoted cell attachment and proliferation on all

layers of Bruch’s membrane, whereastransfectants enhanced adhesion and

proliferation on basal lamina and external collagenous layers.

Conclusions. Overexpression ofandintegrin could enhance ARPE-19 cells adhesion and proliferation on Bruch’smembraneexplant.Modification ofintegrin expression in RPE cells to promote cellular adhesion and proliferation might be a

(5)

Introduction

Aged-related macular degeneration (AMD) is the leading cause of visual impairment

in developed countries in patients over 50 years of age.1, 2Currently, there is no

satisfactory treatment to reverse the visual loss in AMD.3-5Excision of submacular

choroidal new vessels (CNV) combined with retinal pigment epithelial (RPE) cells

transplantation that provides the chance to remove pathological lesions and prevent

secondary choriocapillaris atrophy, has been proposed as a treatment for choroidal

neovascularization.6-8However, little patients gain significant visual improvement

from such surgery9, 10Increasing evidences suggest the inability of transplanted RPE

cells to attach and survive on Bruch’s membrane (BM) is one of the major causes for

RPE transplant failure and poor visual outcomes.11, 12Therefore, to enhance adherence

and repopulation of transplanted RPE cells to Bruch’s membrane became the key step

for the success of RPE transplantation.

RPE transplantation is often performed after surgical removal of subfoveal

neovascualr membranes, which can damage and expose deeper layers of BM, such as

inner collagenous layer, elastic layer and even outer collagenous layer.13, 14Thus, RPE

cells harvested for transplantation usually need to adhere, proliferate and function on

deeper layers of BM. The anatomic layer of BM available for RPE attachment affects

(6)

of the attached cells.15Several in vitro studies demonstrated that the RPE

reattachment rate was highest to the inner aspects of BM and decreased as deeper

layers of BM are exposed.15, 16Zarbin had reported that freshly harvested aged human

RPE cells fail to attach onto the basal lamina or ICL of BM, because they do not

express integrins necessary for attachment.17He found that up-regulation of integrins

by culturing promotes more efficient RPE attachment to and survival on BM. This

finding suggested that selective up-regulation of integrins responsible for adhesion to

BM is a promising strategy to enhance transplanted cells to attach to host BM.

The attachment of RPE cells to Bruch’s membrane is mainly mediated by an

interaction between of integrins on the RPE surface and ligands in the extracellular

matrix, including laminin, fibronectin, vitreonectin and collagen.18, 19Integrins are

heterodimeric molecules composed of a noncovalently boundandsubunit.20-22 There are 18and 8subunits that are known to assemble into 24 distinct integrins.23,

24

. Integrinis a receptor of laminin, the major component of Bruch’s membrane.

25, 26

Integrinis widespread expressed at the basal surface of most epithelial cells and plays an essential role in cell adhesion to the underlying basement membrane.27,

28

.Additionally, integrinalso contributed to proliferation, migration and differentiation of cells.29, 30Previous electron microscopic (EM) studies of the

(7)

junction of RPE and BM revealed that hemidesmosomes was present in the basal

surface of RPE cells, adding in maintaining cohesion between RPE and Bruch’s

membrane.31, 32Since hemidesmosome is mainly composed of integrinthis finding implied that integrinmay also present on the surface of RPE cells. However, the roles of integrinon RPE cells adhesion to extracellular matrix have not been investigated.

In this study, we investigated the expressions and the roles of integrinin

ARPE-19 cells attachment to extracellular matrix. Furthermore, we determined

whether genetically modified RPE cell to overexpress integrinandsubunit could enhance adhesion and proliferation on various layers of Bruch’s membrane.

(8)

Materials and Methods

Antibodies and reagents

Monoclonal antibodies used in this study were: anti-integrin6(clone GoH3),

anti-integrinclone 3E1) and anti-integrinclone 6S6) from Chemicon

(Temecula, CA). Laminin, fibronectin, vitreonectin and collagen type I were obtained

from Sigma-Aldrich (St. Louis, MO).

Cell culture

ARPE-19 cells were purchased from the Bioresource Collection and Research Center

(BCRC, Hsinchu, Taiwan) and cultured in Dulbecco’smodified Eagle’smedium/F-12

human aminiotic membrane nutrient mixture (DMEM/F-12; Sigma-Aldrich, St. Louis,

MO) with 10% fetal bovine serum (FBS; Invitrogen, Carlsbad, CA) in a humidified

incubator at 370C in an atomosphere of 5% CO2.

RNA Extraction and Reverse Transcription-Polymerase Chain Reaction

Total RNA was extracted from ARPE-19 cells with Trizol reagent (Invitrogen-Gibco, Grand Island,NY,USA)according to themanufacturer’sinstructions.Onemicrogram of total RNA from each sample was annealed for 5 min at 65C with 300ng

(9)

U Moloney murine leukemia virus reverse transcriptase (MMLV-RT)

(Invitrogen-Gibco, Grand Island, NY, USA) per 50g reaction for 1 h at 37C. The reaction was stopped by heating for 5 min at 90C.

The cDNA obtained from 0.5 μg totalRNA wasused asatemplateforPCR

amplification. Oligonucleotide primers were designed based on Genbank entries for

human,and β-actin. The following primers were used for amplification

reaction: For, forward primer 5'- GTGTTGCCAACCAGAATGGCTCGC-3′; reverse primer 5'-CAGTCACTCGAACCTGAGTGCCTGC-3’; For, forward primer5′-ACAGGAGGGGTTAAAGCTGC-3′;reverseprimer5′

-GCAGCTTTAACCCCTCCTGT-3′;Forforward primer

5'-CCTCATACTTCGGATTGACC; reverse primer 5’

-TGTTCAGTGCAGAACCTTCA-3; For β-actin,forward primer5′-GAAC

CCTAAGGCCAACCGTG-3′;reverseprimer5′-TGGCATAGAGGTCTTTA CGG-3′.

The amplification was performed in 30 cycles at 55 °C, 30 s; 72 °C, 1 min; 94 °C,

30 s. PCR products were separated by performing gel electrophoresis on 2% agarose

containing ethidium bromide (Sigma, St. Louis, MO, USA) and then analyzed under

ultraviolet light against the DNA molecular length markers. The intensity of the

(10)

Kodak, Rochester, NY, USA), and the amount of PCR-amplifiable material in each

reverse-transcribed sample was standardized against the amount of a housekeeping

gene-actin.

Plasmids and Generation of Point Mutation by Site-directed mutagenesis

Plasmids containing the full-lengthintegrin cDNA (pWT) andintegrin cDNA (p4WT) were gift from Dr. Giancotti. Plasmid p6mut, encoding the mutated S47L

integrin, was generated by polymerase chain reaction (PCR) amplification using the QuickChange site-directed mutagenesis kit (Stratagene, La Jolla, CA), primers

(forward) 5’-AGCCTCTTCGGCTTCTTGCTGGCCATGCAC-3’and (reverse)

5’-GCCAGTGCATGGCCAGCAAGAAGCCGAAGA-3’and plasmid pWT as a template; the primer sequences of plasmid4mutext,encoding the mutated Q155L4

integrin, were: (forward)5’-TCAGCGTCCCGCTGACGGACAT GAGGC-3’and

(reverse)5’-TCATGTCCGTCAGCGGGACGCTGA-3’and plasmid p4WT as a

template; the primer sequences of plasmid4mutint, encoding the mutated R1281W

4integrin, were: (forward)5’-AACCCTAAGAACTGGATCGTTG CTTATTG-3’and

(reverse)5’-CAATAAGCAGCATCCAGTTCTTAGGGTT-3’and plasmid p4WT as

(11)

Transient Transfections

ARPE-19 Cells were seeded in 6-well tissue culture plates at 1.3x 105cells per well

and growth for 48 h in MEM/10% FCS at which time they reach about 90%

confluence. The medium was replaced with serum-free medium, and after 4 hours of incubation the cultures were transfected with 2g of each mutanted construct or full-length integrin6or4cDNA constructs or corresponding empty constructs,

using Lipofectamine 2000 (Invitrogen, Carlsbad, CA)in accordance with the

manufacturer’s instructions. Transfection efficiency was analyzed 24 hours after

transfection by determining the population of cells with GFP expression.

Flow cytometry

Subconfluent cells were washed twice with phosphate-buffer saline (PBS) and

harvested by trypsin/ethylenediamine tetraacetic acid (EDTA) (0.25%wt:vol, 5mM).

Cells were washed once in PBS containing 10% FBS. Cells were incubated with

monoclonal anti-integrin antibodies for 60 minutes at 40C and washed twice with PBS.

Fluorescein isothiocyanate (FITC)-conjugated rabbit anti-rat6 antibody (Jackson

ImmunoResearch laboratories, West Grove, PA, USA) or phycoerythrin

(PE)-conjugated anti-mouse4 secondary antibody (Sigma-Aldrich, St. Louis, MO)

(12)

resuspended in 0.5ml PBS with 10% FBS. Labelled cells were scanned on a

FACSCalibur cytometry (Becton Dickinson) and analysed using CellQuest software.

For each sample the data from 1x104events were collected. All data are presented as

the mean fluorescence intensity, which is proportion to the logarithm of the

fluorescence intensity.

Preparation of different layers of BM

Bruch’s membrane explants were prepared as previously described by Del Priore and

Tezel32. In brief, a full-thickness circumferential incision was made posterior to the

ora serrata and the anterior segment and vitreous were carefully removed. Four radial

incisions were then made and the sclera was peeled away. A circumferential incision

was made into the subretinal space 1 mm posterior to the ora serrata. The

choroids-BM-RPE complex was then carefully peeled toward the optic disc and

removed after trimming its attachment to the optic nerve. Native RPE was removed

by bathing the explant with 0.02N ammonium hydroxide in a 50-mm polystyrene

petri dish (Facon, Becton Dickinson, Lincoln Park, NJ) for 20 minutes at room

temperature, followed by washing 3 times in phosphate-buffered saline (PBS).

(13)

polycarbonate-polyvinylpyrrolidone membrane with 0.4-m pores (Millipore, Bedford, Mass) with the basal lamina facing toward the membrane. The agarose was allowed to solidify at

40C, and the hydrophilic membrane was peeled off along with the basal lamina (BL)

of the RPE, thus exposing the bare internal collagenous layer (ICL). Triplicate buttons

from each eye were further treated at 370C with1-mg/ml collagenase (Sigma-Aldrich,

St. Louis, MO) in PBS at pH 7.5 for 1 hour to remove the internal collagenous layer

and expose the elastic layer (EL) and with collagenase followed with 20 U/ml elastase

(Sigma-Aldrich, St. Louis, MO) in PBS at pH 8.5 for 1 hour to digest the internal

collagenous and elastin layers and expose the external collagenous layer (ECL). After

the enzymatic treatment, 6 mm diameter peripheral buttons were trephined and placed

on 4% agarose in untreated polystyrene 96- well plates. The well were gently rinsed

with PBS 3 times for 5 minutes and then stored at 40C.

RPE reattachment Studies

Plates were pre-coated with the ECM molecules: laminin (10g/ml), fibronectin ( 10 g/ml), vitronectin (10 g/ml ) or collage type IV (25g/ml) in PBS, in a total volume of 100L, for 4 hours at room temperature. Fifteen thousand RPE cells were plated

on 96-well tissue culture plates pre-coating with ECM or different layers of Bruch’s

(14)

penicillin G, 100g/ml streptomycin, 5 g/ml gentamicin, and 2.5 g/ml amphotericin B was added to reach a final volume of 200L in each well. Cells were allowed to

attach to the surface of ECM for 6 hours or Bruch’s membrane for 24 hours in a

humidified atmosphere of 95% air and 5% carbon dioxide at 370C in phenol red-free

MEM. Unattached cells were removed from the tissue culture plates by gently

washing the wells 3 times with MEM

Assay for RPE adhesion

The number of attached live RPE cells in each well was determined with a

colorimetric assay that indirectly estimates the number of live cells by measuring

intracellular dehydrogenase activity (CellTiter 96 Aqueous one solution cell

proliferation assay; Promega, Madison, WI). The dehydrogenase enzymes in live cells

reduce MTS (3-(4,5-dimethyothiazol-2-yl)-5-(3-carboxymethoxyphenyl)

-2-(4-sulfophenyl)-2H-tetrazolium) into formazan in the presence of phenazine

methosulfate (PMS). The quantity of the formazan product can be determined from

the absorbance at 490 nm is directly proportional to the number of living cells in

(15)

Twenty microliters of freshly prepared MTS/ PMS solution (20:1) was added to each

well, resulting in a final concentration of 333 µg/mL of MTS and 25µmol of PMS.

Plates were incubated for 4 hours at 37°C; 100 µL of medium from each well was

transferred to the corresponding wells of another 96-well plate and read at 490nm

using an enzyme-linked immunosorbent assay plate reader. The corrected absorbance

was obtained by subtracting the average optical density reading from triplicate sets of

control sets containing the BM explant on 4% agarose without plated cells. The RPE

adhesion ratio was as follows: cell adhesion (%) = (mean absorbance of cell attached

to wells/ mean absorbance of total number of cells plated) x100. Pilot studies were

performed to verify that there is a linear and reproducible relationship between cell

number and absorbance at 490nm (data not shown).

Assay for RPE Proliferation

Fifteen thousand ARPE-19 cells were planted on different layers of BM as described

above. Cells were maintained for 24 hours in serum-free MEM containing 100 IU/mL penicillin G, 100g/ml streptomycin, 5 g/ml gentamicin, and 2.5 g/ml amphotericin B. At the end of this period, cell proliferation was stimulated by replacing the medium

with MEM supplemented with 15% fetal bovine serum (FBS) and 1 ng/ml

(16)

MTS assay 24 hours after growth stimulation as described above. The proliferation

ratio was the ratio of the number of viable and attached cells 24 hours after growth

stimulation to the initial number of viable and attached cells on a certain explant.

Scanning electron microscopic analysis

Fifteen thousand RPE cells were plated on different layers of Bruch’s membrane

explants, and serum- and phenol-free MEM containing 100 IU/mL penicillin G, 100g/ml streptomycin, 5 g/ml gentamicin, and 2.5 g/ml amphotericin B was added to reach a final volume of 200L in each well. Cells were allowed to attach to

and proliferate on the surface of different layers of Bruch’s membrane for 7 days in a

humidified atmosphere of 95% air and 5% carbon dioxide at 370C. After gently

washing the wells 3 times with MEM, Bruch’s membrane explants were fixed in

modified Karnovsky fixative (2.5% glutaraldehyde and 2% paraformaldehyde in

0.1M cacodylate buffer [pH 7.4]) at 40C overnight. They were then postfixed in 1%

osmium tetroxide in 0.16 M cacodylate buffer (pH 7.4) for 1 hour, stained in 1%

uranyl acetate buffer, and dehydrated in a graded series of ethyl alcohol (30%-100%).

The samples were then critical point dried ( E 3000; Polaron, Watford Hertfordshire,

UK), mounted on aluminum specimen stubs with carbon-conductive tabs grounded

(17)

Polaron). Samples were examined by SEM (model S-4500 FEG; Hitachi, Tokyo,

Japan) at 15kV accelerating voltage and the images recorded (55P/N film; Polaroid

Corp., Cambridge, MA)

Statistical analysis

Student’st-test was used to compare data between two groups. To compare data among three or more groups, one-way analysis of variance (ANOVA) followed by the

Bonferroni test was used. Data are expressed as means ± SEM and P < 0.05 was

(18)

Results:

Expression ofintegrin6 and subunit mRNA as well as protein in ARPE-19 cells

Integrinsubunit and4subunit mRNAs were detectable in ARPE-19 cells (Fig 1A).

Flow cytometric analysis revealed that integrinsubunit and4 subunit proteins

were also presented on the surface of ARPE-19 cells (Fig 1B).

Transient expression of mutant6andin ARPE-19 cells and their effects on

cells adhesion to laminin

To investigate the role of integrin6in RPE cell adhesion to extracellular matrix,

we selectively mutated the amino acid residues of integrinand4, which are

critical for binding function, and then tested the binding ability to laminin. Three

cDNA constructs: each contained single amino acid mutant:mutant by S→L substitution at position Serine 47(mut);mutant by Q→L substitution at position glutamine155 (mutex) andmutant by R→W substitution at position arginine

1281(mutin) were generated by site-directed mutagenesis. Sequencing of the

products confirmed that only the intended mutation was introduced (data not shown).

(19)

surface expression of integrin6subunit andsubunit were determined by flow

cytometry. The expression of integrinsubunit andsubunit in mock-transfectants was similar to that in parental cells. Transfection with themut cDNA construct led to a significant decrease in surface expression of integrinsubunit, but had no effects onsubunit, compared to parental cells. Similarity,mutex-transfectants

exhibited a significant decrease in the expression of integrinsubunit, but no difference in surface expression of integrinsubunit compared to parental cells. However, there was no difference in surface expression of both6andsubunit

betweenmutint-transfectants, and parental cells (Figure 2A).

We next examined the adhesion ofmut-,mutex-,mutin-transfectants,

mock-transfectants and parental cells to laminin. Transfection with themut cDNA andmutexcDNA resulted in a statistically significant reduction in binding to

laminin, compared with mock- transfectants and parental cells, whereas

mutin-transfectants did not display significant differences in ligand binding to

laminin than mock-transfectant and parental cells (Fig 2B).

Effects ofcDNA -transfectants,cDNA -transfectants on adhesion of cells to

(20)

To study whether transfection ofcDNAandcDNAconstruct into ARPE-19

cell could enhance binding to various extracellualr matrixes, we first examined the

expression levels of,and4subunits on the surface ofcDNA-and

cDNAtransfectants by flow cytometry analysis. As shown in Figure 3A,

transfection with thecDNA construct into ARPE-19 cells resulted in overexpression of the integrin6,andsubunit on cell surfaceHowever,

transfection with thecDNAconstruct led to the overepxression of both6 and subunits in the surface of ARPE-19 cells. The level of1 subunit appeared no change incDNAtransfectants.

Next, we performed adhesion assays to determine the adhesion of

cDNAtransfectants andcDNAtransfectants tothe extracellular matrix: laminin (LN), fibronectin (FN), vitreonectin (VN) and collagen type IV (Col IV). The cDNA -transfectants showed a significant increase (P < 0.05) in adhesion to LN and FN compared to mock-transfectants and parental cells. No statistically significant

difference was found in binding ability to Col IV and VN amongcDNA -transfectants, mock-transfectants, and parental cells. In addition,

(21)

to mock-transfectants and parental cells. There were no statistically significant

differences in binding ability to FN, VN and Col IV amongcDNA -transfectants , mock-transfectants, and parental cells (Figure 3B).

Effects ofcDNA -transfectants,cDNA -transfectants on adhesion of cells to

different layers of Bruch’s membrane

To study whether transfection ofcDNAandcDNAconstructs into ARPE-19 cells could enhance adhesion to possible planes of BM after submacular surgery, we

compared the adhesion betweencDNA -transfectants,cDNA -transfectants and mock-transfectants, parental cells to different layers of Bruch’s membrane. The cDNAtransfectants showed a significant increase (P < 0.05) in adhesion to BL, ICL, EL and ECL of BM compared to mock-transfectants and parental cells. In

contrast, thecDNAtransfectants showed a significant increase (P < 0.05) in adhesion to BL and ICL than mock-transfectants and parental cells, but not to EL and

ECL (Figure 4A).

Effects ofcDNA -transfectants,cDNA -transfectants on proliferation rates

(22)

The proliferation rates ofcDNAtransfectants on BL, ICL, EL and ECL of BM were significantly higher than those of mock-transfectants and parental cells. The cDNAtransfectants showed significantly increased proliferation (P < 0.05) on BL, ICL and EL than mock-transfectants and parental cells (Figure 4B).

The ability ofcDNA- andcDNA- transfectants to repopulate different

layers of Bruch’s membrane

To investigate whether transfection with thecDNAandcDNA constructs into ARPE-19 cells could promote proliferation and repopulation to various layers of

Bruch’s membrane, we compared the surface coverage and morphology of

transplanted cells betweencDNA-,cDNA- and mock-transfectants to different layers of Bruch’s membrane after an incubation period of 7 days by scanning electron

microscope (SEM). ThecDNA-transfectants could almost completely cover the explant with BL, ICL, EL and ECL. SEM ofcDNA-transfectants on BL had formed a continuous layer with fairly uniform hexagonal shape (Figure 5A). On ICL, cDNA-transfectants were also flattened and formed a continuous layer, but some gaps and small defects in the monolayer are present (Figure 5B). SEM ofcDNA

(23)

-transfectants on EL showed many defects in Bruch’s membrane coverage.

Resurfacing RPE cells tended to be of variable morphology and flattened. On the

margin of defects, numerous pseudopodia extended from cells to the defects (Figure

5C). On ECL, despite the explants were almost complete coverage, many large RPE

defects were present. Some cells remained round without flattened in the margin of

defects (Figure 5D).

ThecDNA -transfectants could resurface the explants with BL, ICL. Similar to cDNA-transfectants,cDNA-transfectants on BL could form a continuous layer with fairly uniform hexagonal shape (Figure 5E). SEM ofcDNA-transfectants on ICL showed almost complete coverage by flat cells, but many RPE defects several

cell diameters wide were present (Figure 5F). ThecDNA-transfectants could not survival on EL and ECL. SEM showed that no intact cells were present, revealing the

underlying elastic and collagen fiber of Bruch’s membrane bed (Figure 5G, H)

SEM of mock-transfectants on BL showed complete coverage, but some small defects

were present (Figure 5I). However, mock-transfectants could not be survival on ICL,

(24)

0Discussion

RPE cells that remain unattached to a substance after delivery into the subretinal

space may undergo apoptosis, so it is important to optimize the conditions for RPE

reattachment to Bruch’s membrane in order to promote graft survival.35In this study,

we demonstrated that6and4integrin subunits were present on the surface of

ARPE-19 cells, mediating the adhesion function to laminin, the major components of

Bruch’s membrane. Transduction of integrin6orgene into ARPE-19 cells

resulted in overexpression of both integrin6and4on cell surface, with significantly

increased adhesion to laminin. In Bruch’s membrane explant model,and –transfectants also exhibit better adhesion and proliferation on deeper layers of

Bruch’s membrane than mock-transfectants or parental cells. Taken together, these

results indicated that genetically modified ARPE-19 cells that overexpression of integrin could enhance cells adhesion and proliferate on deeper layers of Bruch’s membrane explant. Modification of integrin expression in RPE cells to

promote cellular adhesion and proliferation might be an alternative strategy to

increase the successful rate of RPE transplantation.

In human, pathogenic mutations in either theor thegenes result in blistering

(25)

with pyloric atresia, in which expression of theorintegrin is aberrant.36, 37In transgenic mouse studies, ablation oforintegrin subunits produced a phenotype of severe mucocutaneous fragility, phenotypically similar to human diseases.38These

findings emphasized the importance of theintegrin in the adhesion of epithelial cells to the basement membrane.

Several experimental studies had demonstrated the potential of transplantation into

subretinal space of RPE cells genetically modified to produce desired factors to treat

retinal degenerative diseases such as AMD.39Saigo et al., showed that RPE cells

transduced with brain derived-neurotrophic factor (BDNF) gained the ability to

express this factor in subretinal space without induction of host immunologic

reaction.40Lund et al., showed that transplantation of genetically modified RPE cells

to extend their in vitro lifespan into the subretinal space could support photoreceptors

survival and limit the deterioration of visual function in RCS rats.41In the present

study, we widen the scope of ex vivo gene transfer to RPE cells in adjunctive

molecular treatments for AMD by demonstrating the feasibility of genetically

modified ARPE-19 cells to overexpressintegrin in enhancing cells adhesion and

(26)

Specificity-determining loops (SDL) segment in the I-like domain ofintegrinand propeller domain of integrin were found to play crucial roles in the function of ligand binding.42, 43Tsuruta and coworkers demonstrated that substitution of

glutamine residue for leucine (Q155L) within the SDL ofintegrindecreased the adhesion ofintegrinto laminin in rat bladder epithelila cell lines.44Allegra and coworkers demonstrated that mutation S47L ofpropeller domain of integrin dramatically reduced the expression level ofintegrin in epithelial cells.45In the current study, flow cytometry analysis demonstrated that ARPE-19 cell expressing S47L andQ155L) mutations significantly affect expression of integrinand subunits on the cell surface and thus interfered with binding to laminin. These data suggested that integrinis, at least in part, involved in RPE cell adhesion to laminin. This observation was consistent with the results by Aisenbrye et al, who

pointed out the importance of integrinin the adhesion of RPE to laminin by demonstrating that ARPE-19 cells could synthesize laminins and adhere them through -containing integrins,and

Several studies reported that integrin1 subunit participated in the adhesion of RPE

cells to Bruch’s membrane.47, 48In this study, we found that surface expression of

(27)

-transfectants. The molecular basis for the cross-regulation betweenand1integrin

in-transfectants remained unknown. However, there are several examples of

integrin cross-regulation. Sun et al. have demonstrated that transfection of2 cDNA

into murine mammary carcinoma cell lines result in increased expression of both the and 4 integrin subunits.49

In addition, our results showed the adhesion of -transfectants was better than that of-transfectants in elastic and outer

collagenous layers of Bruch’s membrane explant. Since elastic and outer collagenous

layers are mainly composed of collagen and fibronectin,50which were the ligands of

integrin, we speculated the difference in the behavior between-transfectants and -transfectants in elastic and outer collagenous layers of Bruch’s membrane explant possibly reflected the difference in the expressionintegrin.

In addition to initial attachment, the ability of transplanted RPE cells to proliferate on Bruch’s membrane also has great influence on the success of RPE transplantation. Cell proliferation is known to control by many of the same signaling proteins that play

a role in adhesion and also require a proper interaction of integrin receptors with their

ECM ligands.51, 52Tezel et al., demonstrated that inadequate the number, type or

(28)

study, the induction of integrinandoverexpression in -transduced RPE cells led to not only an increase in cells adhesion, but also cell proliferation on deeper

layers of Bruch’s membrane, indicating

The current study has several limitations. First and the major is that porcine Bruch’s

membrane was used for the preparation of different layers of Bruch’s membrane

explants. Although porcine Bruch’s membranes are approximately close to those of

human with typical pentalaminate structure,55they lack age-related alterations in the

molecular composition and ultrastructure. Additional analysis of whether transfection

ofintegrin subunit into ARPE-19 could enhance attachment to human aged

Bruch’s membrane is now undergoing in our laboratory. Second, coherent to Bruch’s

membrane explant model which lack the influence of overlying neurosensory retina

and choroid, the microenvironment of RPE cells seeded onto BM in tissue culture

may be different from the subretinal space. Despite these limitations, the in vivo

Bruch’s membrane explant system is the useful method to quantitatively evaluate the

behavior of various types or genetically modification of human RPE cells on BM

specimens that may resemble the substrate encountered in vivo duration RPE

(29)

In conclusion, we have shown that ex vivo gene transfer of integrin genes can

increase the adhesion and proliferation of transplanted RPE cells on Bruch’s

membrane explant. The possibility of ex vivo gene transfer to RPE cell may widen the

scope of this procedure to include gene therapy or adjunctive molecular treatment of

(30)

Reference:

1. Klein R, Klein BE, Jensen SC, Meuer SM. The five-year incidence and progression of age-related maculopathy: the Beaver Dam Eye Study. Ophthalmology 1997;104:7-21.

2. West SK. Looking forward to 20/20: a focus on the epidemiology of eye diseases. Epidemiol Rev. 2002;22:64-70.

3. Macular photocoagulation study group. Laser photocoagulation of subfoveal neovascualr lesions in age-related macular degeneration: results of a randomized clinical trial. Arch Ophthalmol. 1991;109:1220-1231.

4. Photodynamic theraoy with verteprofin for age-related macular degeneration. American Acandemy of Ophthalmology. Ophthalmology 2000;107:2314-2317. 5. Krzystolik MG, Afshari MA, Admais AP, et al. Prevention of experimwntal

choroidal neovascularization with intravitreal anti-vascular endothelial growth factor antibody fragment. Arch Ophthalmol. 2002;120:338-346.

6. Del Priore LV, Kaplan HJ, Berger AS. Retinal pigment epithelial transplantation in the management of subfoveal choroidal neovascularization. Semin

Ophthalmol. 1997;12:45–55.

7. Del Priore LV, Kaplan HJ, Horbeck R, Jones Z, Swinn M. Retinal pigment epithelial debridement as a model for the pathogenesis and treatment of macular degeneration. Am J Ophthalmol. 1996;122:629–643.

8. Castellarin AA, Nasir MA, Sugino IK, et al. Progressive presumed

choriocapillaris atrophy after surgery for age-related macular degeneration. Retina. 1998;18:143–149.

9. Algvere PV, Berglin L, Gouras P, Sheng Y. Transplantation of fetal retinal pigment epithelium in age-related macular degeneration with subfoveal neovascularization. Graefe’s Arch Clin Exp Ophthalmol. 1994;232:707-716.

(31)

10. Binder S, Stolba U, Krebs I, et al. Transplantation of autologous retinal pigment epithelium in eyes with foveal neovascularization resulting from age-related macular degeneration: a pilot study. Am J Ophthalmol. 2002;133:215–225. 11. Algvere PV, Gouras P, Dafgard Kopp E. Long-term outcome of RPE allografts

in non-immunosuppressed patients with AMD. Eur J Ophthalmolo. 1999;9:217-230.

12. Del Priore LV, Kaplan HJ, Tezel TH, et al. Retinal pigment epithelial cell transplantation after subfoveal membranectomy in age-related macular degeneration: clinicopathologic correlation. Am J Ophthalmol.

2001;131:472–480.

13. Del Priore LV, Tezel TH, Kaplan HJ. Maculoplasty for age-related macular degeneration: reengineering Bruch’s membrane and human macula. Prog Retin Eye Res 2006;25:539-562.

14. Grossniklaus HE, Hutchinson AK, Capone A, Woolfson J, Lambert HM. Clinicopathologic features of surgically excised choroidal neovascular membranes. Ophthalmology 1994;101:1099-1111.

15. Del Priore LV, Tezel TH, Kaplan HJ. Maculoplasty for age-related macular degeneration: Reengineering Bruch’s membrane and the human macula. Prog. Retinal Eye Res. 2006;25:539-562.

16. Del Priore LV, Tezel TH. Reattachment rate of human retinal pigment epithelium to layers of human Bruch’s membrane. Arch Ophthalmol. 1998;116:335-341.

17. Tezel TH, Del Priore LV, Kaplan HJ. Fate of human retinal pigment epithelial cellsseeded onto layersofhuman Bruch’smembrane.Invest Ophthalmol Vis Sci. 1999;40:467–476.

18. Zarbin MA. Analysis of retinal pigment epithelium integrin expression and adhesion to aged submacular human Bruch’s membrane. Trans Am Ophthalmol Soc 2003;101:499-520.

(32)

19. Avery RL, Glaser BM. Inhibition of retinal pigment epithelial cell attachment by a synthetic peptide derived from the cell-binding domain of fibronectin. Arch Ophthalmol 1986;104:1220-1222.

20. Ho TC, Del Priore LV. Reattachment of cultured human retinal pigment epithelium to extracellularmatrix and human Bruch’smembrane.Invest Ophthalmol Vis Sci. 1997;38:1110–1118.

21. Hynes RO. Integrins:bidirectional, allosteric signaling machines. Cell 2002;110:673-687.

22. van der Flier A, Sonnengerg A.Function and interactions of integrins. Cell Tissue Res 2001;305:285-298.

23. Elner SG, Elner VM. The integrin superfamily and the eye. Invest Ophthalmol Vis Sci 1996;37:696-701.

24. Berman AE, Hozlova NI. Integrins: structure and functions. Membr. Cell Biol. 2000;13:207-244.

25. Sonnenberg A. Integrins and their ligands. Curr. Top Microbiol Immunol. 1993;184:7-35.

26. Kikkawa Y, Sanzen N, Fujiwara H, Sonnenberg A, Sekiguchi K. Integrin binding specificity of laminin-10/11: laminin-10/11 are recognized by andintegrins. J Cell Sci 2000;113:869-876.

27. Niessen CM, Hogervorst F, Jaspars LH, de Melker AA, Delwel GO, Hulsman EHM, Kuikman I, Sonnenberg A. Theintegrin is a receptor for both laminin and kalinin. Exp Cell Res. 1994;211:360-367.

28. Ebihara N, Watanabe Y, Nakayasu K, Kanai A. The expression of laminin-5 and ultrastructure of the interface between basal cells and underlying stroma in the keratoconus cornea. Jpn J Ophthalmol 2001;45:209-215.

29. Sonnenberg A, Calatat J, Jansen H. et al., Integrincomplex is located in hemidesmosomes, suggesting a major role in epidermal cell-basement membrane adhesion. J Cell Biol 1991;113:907-917.

(33)

30. Mercurio AM, Rabinovitz I. Towards a mechanistic understanding of tumor invasion-Lessons from the alpha6beta4 integrin. Semin Cancer Biol 2001; 11:129-141.

31. Mainiero F, Murgia C, Wary KK, et al. The coupling ofintegrin to

Ras-MAPK kinase pathways mediated by Shc controls keratocyte proliferation. EMBO J. 1997;16:2365-2375.

32. Mili H, Bellhorn MB, Henkind P. Specializations of the retinochoroidal juncture. Invest Ophthalmol Vis Sci 1975;14:712-717.

33. Marshall GE, Konstas GP, Reid GG, Edwards JG, Lee WR. Type IV collagen and laminin in Bruch’s membrane and basal linear deposit in the human macula. Br. J Ophthalmol 1992;76:607-614.

34. Stepp MA, Michaud SS, Tsidale A, Elwell J, Gipson IK.integrin heterodimer is a component of hemidesmosomes. Proc Natl Acad Sci USA 1990;87:8970-4.

35. Spinardi L, Einheber S, Cullen T, Milner TA, Giancotti FG. A recombinant tail-less integrinsubunit disrupts hemidesomsomes, but does not suppress mediated cell adhesion to laminin. J Cell Biol 1995;129:473-487. 36. Tezel TH, Del Priore LV, Kaplan HJ. Reattachment to a substrate prevents

apoptosis of human retinal pigment epithelium. Graefes Arch Clin Exp Ophthalmol. 1997;235:41-47.

37. Ruzzi L, Gagnous-Palacios L, Pinola M. et al. A homozygous mutation in the integrin6 gene in junctional epidemolysis bullosa with pyloric atresia. J Clin Invest 1997;99:2826-2831.

38. Ashton GHS, Sorelli P, Mellerio JE, Keane FM, Eady RAJ, McGrath JA.64 integrin abnormalities in junctional epidermolysis bullosa with pyloric atresia. Br J Dermatol 2001;144:408-414.

39. van der Neut R, Krimpenfort P, Calafat J, et al. Epithelial detachment due to absence of hemidesmosomes in integrin4 null mice. Nat Genet 1996;13:366-9.

(34)

40. Binder S, Stanzel BV, Krebs I, Glittenberg C. Transplantation of the RPE in AMD. Prog. Retinal Eye Res. 2007;26:516-54.

41. Saigo Y, Abe T, Hojo M., Tomita H, Sugano E, Tamai M. Transplantation of transduced retinal pigment epithelium in rats. Invest. Ophthalmol. Vis Sci 2004;45:1996-2004.

42. Lund RD, Adamson P, Sauve Y. et al., subretinal transplantation of genetically modified human cell lines attenuates loss of visual function in dystrophic rats. Proc Natl Acad Sci U S A. 2001;14;98:9942-7

43. Takagi J, De Bottis DP, Erickson HP, Springer TA. The role of the specificity-detemining loop of the integrinsubunit I-like domain an autonomous expression, association with thesubunit, and ligand binding. Biochemistry 2002;41:4339-4347.

44. kamata T, Tieu KK, Iric A, Springer TA, Takada Y. Amino acid residues in the apha IIb subunit that are critical for ligand binding to integrin alphaIIbbeta 3 are clustered in the beta-propeller model. J Biol Chem 2001; 276:44275-44283. 45. Tsuruta D, Hopkinson SB, Lane KD, Werner ME, Cryns VL, Jones JCR.

Crucial role of the specificity-determining loop of the integrin4subunit in the

binding of cells to laminin-5 and outside-in signal transduction. J Biol Chem 2003;278:38707-38714.

46. Allergra M, Gagnoux-Palacios L, Gache Y, et al. Rapid decay ofintegrin caused by a mis-sense mutation in the propeller domain results in sever junctional epidermolysis bullosa with pyloric atresia. J. Invest Dermatol 2003;121:1336-1343.

47. Aisenbrey S, Zhang M, Bacher D, Yee J, Brunken WJ, Hunter DD. Retinal pigment epithelial cells synthesize laminins, including laminin 5, and adhere to them through-and 6-containing integrins. Invest Ophthalmol Vis Sci. 2006;47:5537-5544.

(35)

48. Chu P, Grunwald GB. Functional inhibition of retinal pigment epithelial cell-substrate adhesion with a monoclonal antibody against the1 subunit of integrin. Invest Ophthalmol Vis Sci 1991;32:1763-1769

49. Chu P, Grunwald GB. Identification of the 2A10 antigen of retinal pigment epithelium as a1 subunit of integrin. Invest Ophthalmol Vis Sci

1991;32:1757-1762.

50. Sun H, Santoro SA, Zutter MM. Downstream events in mammary gland

morphogenesis mediated by reexpression of the alpha2beta1 integrin: the role of the alpha6 and beta4 integrin subunits. Cancer Res. 1998;58:2224-33.

51. Das A, Frank RN, Zhang NL. et al. Ultrastructural localization of extracellular matrix components in human retinal vessels and Bruch’s membrane. Arch Ophthalmol 1990;108:421-429.

52. Greenwood JA, Murphy-Ullrich JE. Signaling of de-adhesion in celluar regulation and motility. Microsc Res Tech. 1998;43:420-432.

53. Davey G, Buzzai M, Assoian RK. Reduced expression of alpha5beta1 integin prevents spreading-dependent cell proliferatin. J Cell Sci. 1999;112:4663-4672. 54. Tezel TH, Del Priore LV. Repopulation of different layers of host human

Bruch’smembraneby retinalpigment epithelial cell grafts. Invest Ophthalmol Vis Sci. 1999;40:767–774.

55. Tezel TH, Del Priore LV, Kaplan HJ. Reengineering of aged Bruch’s membrane to enhance retinal pigment epithelium repopulation. Invest Ophthalmol Vis Sci 2004;45:3337-3348.

56. Braekevelt CR. Fine structure of the choriocapillaris, Bruch's membrane and retinal epithelium of the cow. Anat Histol Embryol. 1986;15(3):205-14.

(36)

Figure legends

Figure 1. (A). Expression of integrin,andsubunit mRNA in ARPE-19 cells. Integrin mRNA was amplified by RT-PCR using gene-specific primers. PCR products

were elecrophoresed in 1.5% agarose gel and stained with ethidium bromide. Bands

of integrin,andsubunits mRNA were clearly visible in ARPE-19 cells. (B). Flow cytometric analysis of surface-expressed integrinandsubunit in ARPE-19 cells. Histograms depict relative fluorescence intensity (log scale) of negative control

(dot lines) and integrinandsubunit (thick lines).

Figure 2. (A) Flow cytometry analysis of surface-expressed integrinand

subunits in mutant-transfected ARPE-19 cells. Mutatedchain were generated by S ->L substitution at position Ser 47(S47L; mutatedchain were generated by Q ->L substitution at position glutamine 155 ((Q155L)) and R ->W substitution at position arginine 1281(4(R1281W)) . The transfectants were stained with either

an anti-subunit antibody or anti-subunit antibody, and were followed by incubation with an appropriate fluoresceinisothiocyanate (FITC)-conjugated or

phycoerythrin (PE) - conjugated secondary antibody. The cells were analyzed for

(37)

as a function of fluorescence intensity (x axis) and are representative of three separate

experiments. Dot lines represent scan obtained with the isotype control antibody; dash

lines represent scan obtained with mock-transfectants; solid line represent

mutant-ormutant-transfectants. (B) Adhesion of ARPE-19 cells expressing6

ormutants on laminin. Cells were seeded on 96-well plates coated with laminin (10g/ml) for 6 hours. Adhesion was measured using a colorimetric assay that measured intracellular dehydrogenase activity. Data represent the mean ± SEM of

triplicate samples from a representative result of three separate experiments. *P<0.05

compared with mock-transfectants and parentalcells, as determined by Student’s t test.

WT=parentalcells

Figure 3. Cell surface expression of the integrinandsubunits incDNA

transfectants andcDNA transfectants (A). Cells were stained with

fluoresceinisothiocyanate (FITC)-conjugated antibodies against these integrins and

analyzed by FACS analysis. The data are expressed as cell number(y axis) plotted as a

function of fluorescence intensity (x axis) and are representative of three separate

experiments. Control was obtained with isotype control antibody. Thin lines

represented scan obtained with untrasfected cells; dash lines represented scan

(38)

(B). Adhesion ofcDNA transfectants, cDNA transfectants, mock-transfectants,

parentalcells to fibronectin, vitreonectin, laminin and collagen IV. Cells were seeded

on 96-well plates coated with laminin (10g/ml), fibronectin (10 g/ml), vitreonectin

(10g/ml) and collagen IV (25 g/ml) and after 4 hours of incubation, adhesion was

measured using a colorimetric assay that measured intracellular dehydrogenase

activity. Data represent the mean ± SEM of triplicate samples from a representative

result of three separate experiments. *P<0.05 compared with mock-transfectants and

parentalcells, as determined by Student’s t test. Wild= parentalcells. BSA=bovine

serum albumin; LN=laminin; FN=fibronectin; VN=vitreonectin; Col IV= collagen

type IV.

Figure 4. Reattachment rate (A) and proliferation rate (B) ofcDNA transfectants,

cDNA transfectants, mock-transfectants and parentalcells on different layers of

Bruch’s membrane (BM) explants. For reattachment rate, cells were seeded on

different layers of BM explants and allowed to attach for 24 hours. For proliferation

rate, cells that initial attached were cultured for another 24 hours. Plates were washed

and adhesion was quantified using a colorimetric assay that measured intracellular

dehydrogenase activity. Data represent the mean ± SEM of triplicate samples from a

representative result of three separate experiments. *P<0.05 compared with

(39)

parental cells. BL= basal lamina layer; ICL= internal collagenous layer; EL= elastic

layer; ECL= external collagenous layer.

Figure 5. Scanning electron microscopic analysis of cellular morphology and cell

coverage of-transfectants (A, D, G, J),cDNA transfectants (B, E, H, K) and

parental cells (C, F, I, L) seeded to different layers of Bruch’s membrane explants for

7 days. ThecDNA-transfectants could almost completely cover the explant with basal lamina (BL) (A), inner collagenous layer (ICL) (D), elastic layer (EL) (G) and

external collagenous layer (ECL) (J). ThecDNA-transfectants could not survival on EL and ECL. SEM showed that no intact cells were present, revealing the

underlying elastic and collagen fiber of Bruch’s membrane bed (G, H) SEM of

mock-transfectants on BL showed complete coverage, but some small defects were

present. (I). However, mock-transfectants could not be survival on ICL, EL and ECL.

參考文獻

相關文件

 眼睛是心靈之窗,心靈是眼神之源。在眼球後方 感光靈敏的角膜含有 1.37 億個細胞,將收到的信 息傳送至腦部。這些感光細胞,在任何時間均可 同時處理

視導項目 視導細項 分數 自評 得分 填表說明 訪視結果說明

Your problem may be modest, but if it challenges your curiosity and brings into play your inventive faculties, and if you solve it by your own means, you may experience the tension

• 是細胞不正常增生,且這些增生的細胞可

倒傳遞神經網路的演算法使 SPOT 假色影像轉換到 SPOT 自然色影 像。影像的結果。(3)以不同天的 SPOT 假色影像進行網路回想,產 生

當時,許多科學家認為“活”的酵素(如酵母細胞)與“沒 有生命”的胃消化液是不同的東西(生機論),但德國生 理學家Kühne則提出後者是所謂的“酵素”

細胞外液(extracellular fluid;ECF) 約佔

•自內分泌系統分泌的激素利用血液輸送到 目 標器官。類固醇和甲狀腺激素是非極 性,且為脂 溶性,所以能通過細胞膜進入 目標細胞。大部分